Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Viruses ; 16(2)2024 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-38400054

RESUMEN

Orthohantaviruses may cause hemorrhagic fever with renal syndrome or hantavirus cardiopulmonary syndrome. Andes virus (ANDV) is the only orthohantavirus associated with human-human transmission. Therefore, emergency vaccination would be a valuable public health measure to combat ANDV-derived infection clusters. Here, we utilized a promising vesicular stomatitis virus (VSV)-based vaccine to advance the approach for emergency applications. We compared monovalent and bivalent VSV vectors containing the Ebola virus (EBOV), glycoprotein (GP), and ANDV glycoprotein precursor (GPC) for protective efficacy in pre-, peri- and post-exposure immunization by the intraperitoneal and intranasal routes. Inclusion of the EBOV GP was based on its favorable immune cell targeting and the strong innate responses elicited by the VSV-EBOV vaccine. Our data indicates no difference of ANDV GPC expressing VSV vectors in pre-exposure immunization independent of route, but a potential benefit of the bivalent VSVs following peri- and post-exposure intraperitoneal vaccination.


Asunto(s)
Vacunas contra el Virus del Ébola , Ebolavirus , Orthohantavirus , Cricetinae , Animales , Humanos , Vesiculovirus/genética , Virus de la Estomatitis Vesicular Indiana/genética , Ebolavirus/genética , Glicoproteínas , Anticuerpos Antivirales
2.
PNAS Nexus ; 1(3): pgac114, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35967978

RESUMEN

Little is known about the temporal patterns of infection and transmission of Lassa virus (LASV) within its natural reservoir (Mastomys natalensis). Here, we characterize infection dynamics and transmissibility of a LASV isolate (Soromba-R) in adult lab-reared M. natalensis originating from Mali. The lab-reared M. natalenesis proved to be highly susceptible to LASV isolates from geographically distinct regions of West Africa via multiple routes of exposure, with 50% infectious doses of < 1 TCID50. Postinoculation, LASV Soromba-R established a systemic infection with no signs of clinical disease. Viral RNA was detected in all nine tissues examined with peak concentrations detected between days 7 and 14 postinfection within most organs. There was an overall trend toward clearance of virus within 40 days of infection in most organs. The exception is lung specimens, which retained positivity throughout the course of the 85-day study. Direct (contact) and indirect (fomite) transmission experiments demonstrated 40% of experimentally infected M. natalensis were capable of transmitting LASV to naïve animals, with peak transmissibility occurring between 28 and 42 days post-inoculation. No differences in patterns of infection or transmission were noted between male and female experimentally infected rodents. Adult lab-reared M. natalensis are highly susceptible to genetically distinct LASV strains developing a temporary asymptomatic infection associated with virus shedding resulting in contact and fomite transmission within a cohort.

3.
Viruses ; 11(3)2019 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-30909389

RESUMEN

Syrian hamsters (Mesocricetus auratus) are a pathogenesis model for the Nipah virus (NiV), and we sought to determine if they are also susceptible to the Cedar virus (CedPV). Following intranasal inoculation with CedPV, virus replication occurred in the lungs and spleens of infected hamsters, a neutralizing antibody was produced in some hamsters within 8 days post-challenge, and no conspicuous signs of disease occurred. CedPV replicated to a similar magnitude as NiV-Bangladesh in type I IFN-deficient BHK-21 Syrian hamster fibroblasts but replicated 4 logs lower in type I IFN-competent primary Syrian hamster and human pulmonary endothelial cells, a principal target of henipaviruses. The coinfection of these cells with CedPV and NiV failed to rescue CedPV titers and did not diminish NiV titers, suggesting the replication machinery is virus-specific. Type I IFN response transcripts Ifna7, Ddx58, Stat1, Stat2, Ccl5, Cxcl10, Isg20, Irf7, and Iigp1 were all significantly elevated in CedPV-infected hamster endothelial cells, whereas Ifna7 and Iigp1 expression were significantly repressed during NiV infection. These results are consistent with the hypothesis that CedPV's inability to counter the host type I IFN response may, in part, contribute to its lack of pathogenicity. Because NiV causes a fatal disease in Syrian hamsters with similarities to human disease, this model will provide valuable information about the pathogenic mechanisms of henipaviruses.


Asunto(s)
Infecciones por Henipavirus/inmunología , Interacciones Huésped-Patógeno/inmunología , Inmunidad Innata , Replicación Viral , Animales , Coinfección/inmunología , Coinfección/virología , Cricetinae , Células Endoteliales/inmunología , Células Endoteliales/virología , Femenino , Henipavirus/patogenicidad , Henipavirus/fisiología , Humanos , Pulmón/virología , Virus Nipah/patogenicidad , Virus Nipah/fisiología , Bazo/virología
4.
Genome Med ; 10(1): 58, 2018 08 07.
Artículo en Inglés | MEDLINE | ID: mdl-30081931

RESUMEN

BACKGROUND: The 2014-2016 Ebola virus (EBOV) outbreak in West Africa highlighted the need for improved therapeutic options against this virus. Approaches targeting host factors/pathways essential for the virus are advantageous because they can potentially target a wide range of viruses, including newly emerging ones and because the development of resistance is less likely than when targeting the virus directly. However, systematic approaches for screening host factors important for EBOV have been hampered by the necessity to work with this virus at biosafety level 4 (BSL4). METHODS: In order to identify host factors involved in the EBOV life cycle, we performed a genome-wide siRNA screen comprising 64,755 individual siRNAs against 21,566 human genes to assess their activity in EBOV genome replication and transcription. As a screening platform, we used reverse genetics-based life cycle modelling systems that recapitulate these processes without the need for a BSL4 laboratory. RESULTS: Among others, we identified the de novo pyrimidine synthesis pathway as an essential host pathway for EBOV genome replication and transcription, and confirmed this using infectious EBOV under BSL4 conditions. An FDA-approved drug targeting this pathway showed antiviral activity against infectious EBOV, as well as other non-segmented negative-sense RNA viruses. CONCLUSIONS: This study provides a minable data set for every human gene regarding its role in EBOV genome replication and transcription, shows that an FDA-approved drug targeting one of the identified pathways is highly efficacious in vitro, and demonstrates the power of life cycle modelling systems for conducting genome-wide host factor screens for BSL4 viruses.


Asunto(s)
Antivirales/farmacología , Ebolavirus/fisiología , Genoma Humano , Replicación Viral , Animales , Línea Celular Tumoral , Chlorocebus aethiops , Clonación Molecular , Ebolavirus/efectos de los fármacos , Ebolavirus/patogenicidad , Técnicas de Silenciamiento del Gen , Células HEK293 , Interacciones Huésped-Patógeno/genética , Humanos , Células Vero
5.
J Infect Dis ; 218(suppl_5): S409-S417, 2018 11 22.
Artículo en Inglés | MEDLINE | ID: mdl-30085162

RESUMEN

Ebola virus (EBOV) and Marburg virus (MARV) outbreaks are highly lethal, and infection results in a hemorrhagic fever with complex etiology. These zoonotic viruses dysregulate the immune system to cause disease, in part by replicating within myeloid cells that would normally innately control viral infection and shape the adaptive immune response. We used triple knockout (TKO)-bone marrow, liver, thymus (BLT) humanized mice to recapitulate the early in vivo human immune response to filovirus infection. Disease severity in TKO-BLT mice was dissimilar between EBOV and MARV with greater severity observed during EBOV infection. Disease severity was related to increased Kupffer cell infection in the liver, higher levels of myeloid dysfunction, and skewing of macrophage subtypes in EBOV compared with MARV-infected mice. Overall, the TKO-BLT model provided a practical in vivo platform to study the human immune response to filovirus infection and generated a better understanding of how these viruses modulate specific components of the immune system.


Asunto(s)
Médula Ósea/virología , Ebolavirus/patogenicidad , Marburgvirus/patogenicidad , Células Mieloides/virología , Timo/virología , Animales , Médula Ósea/inmunología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/virología , Inmunidad/inmunología , Hígado/inmunología , Hígado/virología , Macrófagos/inmunología , Macrófagos/virología , Enfermedad del Virus de Marburg/inmunología , Enfermedad del Virus de Marburg/virología , Marburgvirus/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Mieloides/inmunología , Timo/inmunología , Virulencia/inmunología
6.
Emerg Infect Dis ; 24(9): 1696-1699, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29882740

RESUMEN

Lassa virus, the cause of Lassa fever in humans, is endemic to West Africa. Treatment of Lassa fever is primarily supportive, although ribavirin has shown limited efficacy if administered early during infection. We tested favipiravir in Lassa virus-viremic macaques and found that 300 mg/kg daily for 2 weeks successfully treated infection.


Asunto(s)
Amidas/uso terapéutico , Antivirales/uso terapéutico , Fiebre de Lassa/veterinaria , Virus Lassa/aislamiento & purificación , Macaca , Enfermedades de los Monos/tratamiento farmacológico , Pirazinas/uso terapéutico , Amidas/administración & dosificación , Animales , Antivirales/administración & dosificación , Femenino , Inyecciones Subcutáneas/veterinaria , Fiebre de Lassa/tratamiento farmacológico , Pirazinas/administración & dosificación , Distribución Aleatoria , Resultado del Tratamiento
7.
Cell Rep ; 23(6): 1806-1816, 2018 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-29742435

RESUMEN

Ebola virus (EBOV), isolate Makona, the causative agent of the West African EBOV epidemic, has been the subject of numerous investigations to determine the genetic diversity and its potential implication for virus biology, pathogenicity, and transmissibility. Despite various mutations that have emerged over time through multiple human-to-human transmission chains, their biological relevance remains questionable. Recently, mutations in the glycoprotein GP and polymerase L, which emerged and stabilized early during the outbreak, have been associated with improved viral fitness in cell culture. Here, we infected mice and rhesus macaques with EBOV-Makona isolates carrying or lacking those mutations. Surprisingly, all isolates behaved very similarly independent of the genotype, causing severe or lethal disease in mice and macaques, respectively. Likewise, we could not detect any evidence for differences in virus shedding. Thus, no specific biological phenotype could be associated with these EBOV-Makona mutations in two animal models.


Asunto(s)
Ebolavirus/genética , Ebolavirus/patogenicidad , Genoma Viral , Mutación/genética , Animales , Secuencia de Bases , Coagulación Sanguínea , Modelos Animales de Enfermedad , Femenino , Fiebre Hemorrágica Ebola/sangre , Fiebre Hemorrágica Ebola/virología , Macaca mulatta , Masculino , Ratones , Ratones Noqueados , Filogenia , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/metabolismo , Análisis de Supervivencia , Carga Viral , Viremia
8.
Artículo en Inglés | MEDLINE | ID: mdl-29696134

RESUMEN

The common small animal disease models for Zika virus (ZIKV) are mice lacking the interferon responses, but infection of interferon receptor α/ß knock out (IFNAR-/-) mice is not uniformly lethal particularly in older animals. Here we sought to advance this model in regard to lethality for future countermeasure efficacy testing against more recent ZIKV strains from the Asian lineage, preferably the American sublineage. We first infected IFNAR-/- mice subcutaneously with the contemporary ZIKV-Paraiba strain resulting in predominantly neurological disease with ~50% lethality. Infection with ZIKV-Paraiba by different routes established a uniformly lethal model only in young mice (4-week old) upon intraperitoneal infection. However, intraperitoneal inoculation of ZIKV-French Polynesia resulted in uniform lethality in older IFNAR-/- mice (10-12-weeks old). In conclusion, we have established uniformly lethal mouse disease models for efficacy testing of antivirals and vaccines against recent ZIKV strains representing the Asian lineage.


Asunto(s)
Modelos Animales de Enfermedad , Receptor de Interferón alfa y beta/genética , Infección por el Virus Zika/mortalidad , Infección por el Virus Zika/patología , Virus Zika/aislamiento & purificación , Aedes , Factores de Edad , Animales , Línea Celular , Chlorocebus aethiops , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Vero , Virus Zika/patogenicidad , Infección por el Virus Zika/virología
9.
J Infect Dis ; 217(1): 58-63, 2017 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-29087482

RESUMEN

Both Ebola virus (EBOV) and Reston virus (RESTV) cause disease in nonhuman primates, yet only EBOV causes disease in humans. To investigate differences in viral pathogenicity, humanized mice (hu-NSG-SGM3) were inoculated with EBOV or RESTV. Consistent with differences in disease in human infection, pronounced weight loss and markers of hepatic damage and disease were observed exclusively in EBOV-infected mice. These abnormalities were associated with significantly higher EBOV replication in the liver but not in the spleen, suggesting that in this model, efficiency of viral replication in select tissues early in infection may contribute to differences in viral pathogenicity.


Asunto(s)
Ebolavirus/crecimiento & desarrollo , Fiebre Hemorrágica Ebola/virología , Hígado/virología , Replicación Viral , Animales , Peso Corporal , Modelos Animales de Enfermedad , Fiebre Hemorrágica Ebola/patología , Humanos , Pruebas de Función Hepática , Ratones , Ratones SCID
10.
J Vet Med Sci ; 79(8): 1453-1460, 2017 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-28674309

RESUMEN

The pandemic 2009 H1N1 influenza A virus emerged in humans and caused the first influenza pandemic of the 21st century. Mexican isolates, A/Mexico/4108/2009 (H1N1) (Mex4108) and A/Mexico/InDRE4478/2009 (H1N1) (Mex4487) derived from a mild case and from a cluster of severe cases, showed heterogeneity in virulence in a cynomolgus macaque model. To compare the more pathogenic differences, we generated recombinant viruses and compared their virulence in ferrets. Ferrets infected with recombinant Mex4487 displayed a slightly higher rate of viral replication and severe pneumonia in the early stage of infection. In contrast, prolonged lower virus shedding of recombinant Mex4108 than that of recombinant Mex4487 was detected in throat swabs. Thus, Mex4487 induces severe pneumonia in infected individuals, whereas Mex4108 might have wide-spreading potential with mild disease.


Asunto(s)
Hurones , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Gripe Humana/virología , Infecciones por Orthomyxoviridae/veterinaria , Pandemias , Animales , Línea Celular , Efecto Citopatogénico Viral , Perros , Femenino , Humanos , Subtipo H1N1 del Virus de la Influenza A/genética , Gripe Humana/epidemiología , Riñón/citología , Masculino , Infecciones por Orthomyxoviridae/virología , Virus Reordenados
11.
Emerg Infect Dis ; 23(6): 985-988, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28318484

RESUMEN

We tested the suitability of the domestic pig as a model for Middle East respiratory syndrome coronavirus (MERS-CoV) infection. Inoculation did not cause disease, but a low level of virus replication, shedding, and seroconversion were observed. Pigs do not recapitulate human MERS-CoV and are unlikely to constitute a reservoir in nature.


Asunto(s)
Infecciones por Coronavirus/veterinaria , Reservorios de Enfermedades/veterinaria , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Enfermedades de los Porcinos/virología , Animales , Infecciones por Coronavirus/virología , Reservorios de Enfermedades/virología , Especificidad del Huésped , Porcinos
12.
Virus Res ; 228: 7-13, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-27840112

RESUMEN

Currently, there is no registered treatment for infections with emerging zoonotic coronaviruses like SARS- and MERS-coronavirus. We here report that in cultured cells low-micromolar concentrations of alisporivir, a non-immunosuppressive cyclosporin A-analog, inhibit the replication of four different coronaviruses, including MERS- and SARS-coronavirus. Ribavirin was found to further potentiate the antiviral effect of alisporivir in these cell culture-based infection models, but this combination treatment was unable to improve the outcome of SARS-CoV infection in a mouse model. Nevertheless, our data provide a basis to further explore the potential of Cyp inhibitors as host-directed, broad-spectrum inhibitors of coronavirus replication.


Asunto(s)
Ciclosporina/farmacología , Coronavirus del Síndrome Respiratorio de Oriente Medio/efectos de los fármacos , Coronavirus del Síndrome Respiratorio de Oriente Medio/fisiología , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/efectos de los fármacos , Coronavirus Relacionado al Síndrome Respiratorio Agudo Severo/fisiología , Replicación Viral/efectos de los fármacos , Animales , Antivirales/farmacología , Línea Celular , Células Cultivadas , Infecciones por Coronavirus/tratamiento farmacológico , Infecciones por Coronavirus/virología , Efecto Citopatogénico Viral/efectos de los fármacos , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Humanos , Ratones , Síndrome Respiratorio Agudo Grave/tratamiento farmacológico , Síndrome Respiratorio Agudo Grave/virología
13.
J Infect Dis ; 214(suppl 3): S308-S318, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27601621

RESUMEN

The study of Ebola virus (EBOV) pathogenesis in vivo has been limited to nonhuman primate models or use of an adapted virus to cause disease in rodent models. Herein we describe wild-type EBOV (Makona variant) infection of mice engrafted with human hematopoietic CD34+ stem cells (Hu-NSG™-SGM3 mice; hereafter referred to as SGM3 HuMice). SGM3 HuMice support increased development of myeloid immune cells, which are primary EBOV targets. In SGM3 HuMice, EBOV replicated to high levels, and disease was observed following either intraperitoneal or intramuscular inoculation. Despite the high levels of viral antigen and inflammatory cell infiltration in the liver, the characteristic histopathology of Ebola virus disease was not observed, and this absence of severe immunopathology may have contributed to the recovery and survival of some of the animals. Future investigations into the underlying mechanisms of the atypical disease presentation in SGM3 HuMice will provide additional insights into the immunopathogenesis of severe EBOV disease.


Asunto(s)
Antígenos Virales/inmunología , Ebolavirus/fisiología , Fiebre Hemorrágica Ebola/virología , Animales , Modelos Animales de Enfermedad , Ebolavirus/inmunología , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/patología , Células Madre Hematopoyéticas/virología , Fiebre Hemorrágica Ebola/inmunología , Fiebre Hemorrágica Ebola/patología , Humanos , Hígado/inmunología , Hígado/patología , Hígado/virología , Linfocitos/patología , Linfocitos/virología , Ratones , Ratones Transgénicos , Células Mieloides/inmunología , Células Mieloides/patología , Células Mieloides/virología , Bazo/inmunología , Bazo/patología , Bazo/virología , Transgenes , Replicación Viral
14.
J Infect Dis ; 214(suppl 3): S355-S359, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27511894

RESUMEN

Antiviral therapeutics with existing clinical safety profiles would be highly desirable in an outbreak situation, such as the 2013-2016 emergence of Ebola virus (EBOV) in West Africa. Although, the World Health Organization declared the end of the outbreak early 2016, sporadic cases of EBOV infection have since been reported. Alisporivir is the most clinically advanced broad-spectrum antiviral that functions by targeting a host protein, cyclophilin A (CypA). A modest antiviral effect of alisporivir against contemporary (Makona) but not historical (Mayinga) EBOV strains was observed in tissue culture. However, this effect was not comparable to observations for an alisporivir-susceptible virus, the flavivirus tick-borne encephalitis virus. Thus, EBOV does not depend on (CypA) for replication, in contrast to many other viruses pathogenic to humans.


Asunto(s)
Antivirales/uso terapéutico , Ciclosporina/uso terapéutico , Brotes de Enfermedades , Ebolavirus/efectos de los fármacos , Fiebre Hemorrágica Ebola/tratamiento farmacológico , África Occidental/epidemiología , Fiebre Hemorrágica Ebola/virología , Humanos , Replicación Viral
15.
Clin Infect Dis ; 63(8): 1026-33, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27531847

RESUMEN

BACKGROUND: The ongoing Ebola outbreak in West Africa has resulted in 28 646 suspected, probable, and confirmed Ebola virus infections. Nevertheless, malaria remains a large public health burden in the region affected by the outbreak. A joint Centers for Disease Control and Prevention/National Institutes of Health diagnostic laboratory was established in Monrovia, Liberia, in August 2014, to provide laboratory diagnostics for Ebola virus. METHODS: All blood samples from suspected Ebola virus-infected patients admitted to the Médecins Sans Frontières ELWA3 Ebola treatment unit in Monrovia were tested by quantitative real-time polymerase chain reaction for the presence of Ebola virus and Plasmodium species RNA. Clinical outcome in laboratory-confirmed Ebola virus-infected patients was analyzed as a function of age, sex, Ebola viremia, and Plasmodium species parasitemia. RESULTS: The case fatality rate of 1182 patients with laboratory-confirmed Ebola virus infections was 52%. The probability of surviving decreased with increasing age and decreased with increasing Ebola viral load. Ebola virus-infected patients were 20% more likely to survive when Plasmodium species parasitemia was detected, even after controlling for Ebola viral load and age; those with the highest levels of parasitemia had a survival rate of 83%. This effect was independent of treatment with antimalarials, as this was provided to all patients. Moreover, treatment with antimalarials did not affect survival in the Ebola virus mouse model. CONCLUSIONS: Plasmodium species parasitemia is associated with an increase in the probability of surviving Ebola virus infection. More research is needed to understand the molecular mechanism underlying this remarkable phenomenon and translate it into treatment options for Ebola virus infection.


Asunto(s)
Coinfección , Ebolavirus , Fiebre Hemorrágica Ebola/complicaciones , Fiebre Hemorrágica Ebola/mortalidad , Malaria/complicaciones , Malaria/parasitología , Parasitemia , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Animales , Niño , Preescolar , Modelos Animales de Enfermedad , Ebolavirus/genética , Femenino , Fiebre Hemorrágica Ebola/diagnóstico , Fiebre Hemorrágica Ebola/epidemiología , Humanos , Lactante , Recién Nacido , Malaria/diagnóstico , Malaria/epidemiología , Masculino , Ratones , Persona de Mediana Edad , Carga de Parásitos , Plasmodium/genética , Tasa de Supervivencia , Carga Viral , Adulto Joven
16.
J Infect Dis ; 214(suppl 3): S360-S366, 2016 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-27496978

RESUMEN

The Ebola virus (EBOV) epidemic in West Africa increased the focus on vaccine development against this hemorrhagic fever-causing pathogen, and as a consequence human clinical trials for a few selected platforms were accelerated. One of these vaccines is vesicular stomatitis virus (VSV)-EBOV, also known as rVSV-ZEBOV, a fast-acting vaccine against EBOV and so far the only vaccine with reported efficacy against EBOV infections in humans in phase III clinical trials. In this study, we analyzed the potential of VSV-EBOV for postexposure treatment of rhesus macaques infected with EBOV-Makona. We treated groups of animals with 1 dose of VSV-EBOV either in a single injection at 1 or 24 hours after EBOV exposure or with 2 injections, half the dose at each time point; 1 control group received the same dose of the VSV-based Marburg virus vaccine at both time points; another group remained untreated. Although all untreated animals succumbed to EBOV infection, 33%-67% of the animals in each treatment group survived the infection, including the group treated with the VSV-based Marburg virus vaccine. This result suggests that protection from postexposure vaccination may be antigen unspecific and due rather to an early activation of the innate immune system. In conclusion, VSV-EBOV remains a potent and fast-acting prophylactic vaccine but demonstrates only limited efficacy in postexposure treatment.


Asunto(s)
Anticuerpos Antivirales/inmunología , Vacunas contra el Virus del Ébola/inmunología , Ebolavirus/inmunología , Fiebre Hemorrágica Ebola/prevención & control , Vacunación , Estomatitis Vesicular/prevención & control , Vesiculovirus/inmunología , África Occidental/epidemiología , Animales , Femenino , Fiebre Hemorrágica Ebola/virología , Humanos , Macaca mulatta , Masculino , Marburgvirus/inmunología , Estomatitis Vesicular/virología
17.
Emerg Infect Dis ; 22(2): 331-4, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26812583

RESUMEN

Rapid sequencing of RNA/DNA from pathogen samples obtained during disease outbreaks provides critical scientific and public health information. However, challenges exist for exporting samples to laboratories or establishing conventional sequencers in remote outbreak regions. We successfully used a novel, pocket-sized nanopore sequencer at a field diagnostic laboratory in Liberia during the current Ebola virus outbreak.


Asunto(s)
Ebolavirus/genética , Fiebre Hemorrágica Ebola/microbiología , Nanoporos , Análisis de Secuencia de ADN/métodos , Brotes de Enfermedades , Genoma Viral , Fiebre Hemorrágica Ebola/epidemiología , Humanos , Mutación
19.
Sci Rep ; 5: 14775, 2015 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-26456301

RESUMEN

With up to 500,000 infections annually, Lassa virus (LASV), the cause of Lassa fever, is one of the most prevalent etiological agents of viral hemorrhagic fever (VHF) in humans. LASV is endemic in several West African countries with sporadic cases and prolonged outbreaks observed most commonly in Sierra Leone, Liberia, Guinea and Nigeria. Additionally several cases of Lassa fever have been imported into North America, Europe and Asia making LASV a global threat to public health. Despite this, currently no approved therapeutic or vaccine exists to treat or prevent LASV infections. Here, using a passaged strain of LASV that is uniformly lethal in Hartley guinea pigs, we demonstrate that favipiravir, a broad-spectrum antiviral agent and leading treatment option for influenza, has potent activity against LASV infection. In this model, once daily treatment with favipiravir significantly reduced viral titers in tissue samples and reduced mortality rates when compared with animals receiving vehicle-only or ribavirin, the current standard of care for Lassa fever. Favipiravir remained highly effective against lethal LASV infection when treatments were initiated nine days post-infection, a time when animals were demonstrating advanced signs of disease. These results support the further preclinical evaluation of favipiravir for Lassa fever and other VHFs.


Asunto(s)
Amidas/farmacología , Anticuerpos Antivirales/biosíntesis , Antivirales/farmacología , Inmunoglobulina G/biosíntesis , Fiebre de Lassa/tratamiento farmacológico , Virus Lassa/efectos de los fármacos , Pirazinas/farmacología , Animales , Animales no Consanguíneos , Anticuerpos Antivirales/sangre , Esquema de Medicación , Cálculo de Dosificación de Drogas , Cobayas , Humanos , Inmunoglobulina G/sangre , Fiebre de Lassa/inmunología , Fiebre de Lassa/mortalidad , Fiebre de Lassa/virología , Virus Lassa/patogenicidad , Virus Lassa/fisiología , Hígado/efectos de los fármacos , Hígado/virología , Pulmón/efectos de los fármacos , Pulmón/virología , Masculino , Ribavirina/farmacología , Bazo/efectos de los fármacos , Bazo/patología , Bazo/virología , Análisis de Supervivencia , Carga Viral/efectos de los fármacos
20.
Vaccine ; 33(24): 2823-9, 2015 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-25865472

RESUMEN

Nipah virus is a zoonotic paramyxovirus that causes severe respiratory and/or encephalitic disease in humans, often resulting in death. It is transmitted from pteropus fruit bats, which serve as the natural reservoir of the virus, and outbreaks occur on an almost annual basis in Bangladesh or India. Outbreaks are small and sporadic, and several cases of human-to-human transmission have been documented as an important feature of the epidemiology of Nipah virus disease. There are no approved countermeasures to combat infection and medical intervention is supportive. We recently generated a recombinant replication-competent vesicular stomatitis virus-based vaccine that encodes a Nipah virus glycoprotein as an antigen and is highly efficacious in the hamster model of Nipah virus disease. Herein, we show that this vaccine protects African green monkeys, a well-characterized model of Nipah virus disease, from disease one month after a single intramuscular administration of the vaccine. Vaccination resulted in a rapid and strong virus-specific immune response which inhibited virus shedding and replication. This vaccine platform provides a rapid means to afford protection from Nipah virus in an outbreak situation.


Asunto(s)
Infecciones por Henipavirus/prevención & control , Virus Nipah/inmunología , Vacunas de ADN/administración & dosificación , Vesiculovirus/inmunología , Vacunas Virales/administración & dosificación , Animales , Chlorocebus aethiops , Cricetinae , Modelos Animales de Enfermedad , Femenino , Glicoproteínas/administración & dosificación , Glicoproteínas/genética , Glicoproteínas/inmunología , Infecciones por Henipavirus/inmunología , Infecciones por Henipavirus/virología , Inmunidad Celular , Masculino , Virus Nipah/genética , Virus Nipah/fisiología , Vacunación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Vacunas de ADN/inmunología , Vesiculovirus/genética , Proteínas del Envoltorio Viral/inmunología , Carga Viral , Vacunas Virales/inmunología , Viremia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...